Knockdown of autophagy-related gene LC3 enhances the sensitivity of HepG2 cells to epirubicin

  • Authors:
    • Wanxin Peng
    • Tong Du
    • Zihao Zhang
    • Fengyi Du
    • Jie Jin
    • Aihua Gong
  • View Affiliations

  • Published online on: February 5, 2015     https://doi.org/10.3892/etm.2015.2266
  • Pages: 1271-1276
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hepatocellular carcinoma (HCC) is a major public health problem. Despite new chemotherapeutic treatments, drug resistance remains a major clinical obstacle to successful treatment in HCC patients. Therefore, novel therapeutic targets and new modalities of treatment are urgently required. In this study, tetracycline‑inducible lentivirus‑mediated RNA interference (RNAi) was employed to knock down microtubule‑associated protein 1 light chain 3 (LC3) gene, which encodes a key protein in the induction of autophagy, to study the protective function of autophagy in liver cancer tolerant to epirubicin. The effect of combined treatment with lentiviral shLC3 and epirubicin on cell growth and chemosensitivity to epirubicin in the HCC cell line HepG2 were also investigated. The results demonstrated that lentivirus‑mediated LC3 silencing significantly inhibited cell proliferation. In addition, combined treatment with lentiviral shLC3 and epirubicin significantly decreased the survival rate of HepG2 cells, compared with that following treatment with either agent alone. Overall, the results from this study suggest for the first time, to the best of our knowledge, that LC3 plays a key role in HCC tumorigenesis, and is a novel therapeutic target for HCC.

Introduction

Hepatocellular carcinoma (HCC) is the fifth most prevalent type of cancer and the third most common cause of cancer-associated mortality worldwide. Each year there are ~630,000 new cases of HCC, more than half of which occur in China (1). Despite recent advances in the understanding of the molecular basis of HCC and new therapeutic approaches, the mortality rate has declined only modestly. Drug resistance remains a major clinical obstacle to successful treatment in HCC patients (2).

Autophagy (which means self-eating) is a dynamic process in which subcellular membranes are rearranged to sequester cytoplasmic constituents, including organelles, for delivery to a lysosome or vacuole where the sequestered cargo is degraded and recycled (3). The formation of autophagosomes, double-membrane vesicles that deliver the cytoplasmic material, is central to this process. Autophagosome formation involves the conjugation of cytosolic microtubule-associated protein light chain 3 (LC3-I) with phosphatidylethanolamine to form LC3-phosphatidylethanolamine (LC3-II) as an essential process. Therefore, the ratio of LC3-II and LC3-I levels can be used as a marker to reflect the activation of autophagy (4,5).

During periods of nutrient deprivation, autophagy degrades intracellular proteins to serve as substrates for ATP generation (6). Autophagy is essential during starvation, cellular differentiation, cell death and aging, and also in the prevention of certain types of cancer and increased tumor cell survival (7,8). Autophagy induction (autophagic activity above basal levels) may occur with anticancer drug treatment as an adaptive response and can lead to chemoresistance (9). For example, when exposed to low doses of radiation, breast, colon and prostate carcinoma cells accumulate acidic vesicular organelles (AVOs) due to autophagy; this is a defense mechanism that increases the survival of irradiated cells (10). The p53-induced transcription of proteins involved in the positive regulation of the autophagy pathway is the common mechanism by which different types of anticancer drugs such as DNA-damaging agents, microtubule interfering molecules, and kinase inhibitors trigger autophagy (11,12).

Defects of autophagy are associated with numerous diseases and tumors (13,14). However, only a few studies have examined hepatocellular carcinoma (HCC) in relation to these processes (15,16). Therefore, in the present study, inducible lentivirus-mediated RNA interference (RNAi) of LC3 was used to investigate the association between autophagy and chemoresistance in HCC. The aim of the study was to improve our understanding of autophagy in human liver cancers and delineate the possible role of autophagy as a novel target for anticancer therapy.

Materials and methods

Cell culture and treatment conditions

The human hepatoma cell line HepG2 was purchased from the Cancer Cell Repository (Shanghai Cell Bank, Shanghai, China) and maintained in Dulbecco’s modified Eagle’s medium (high glucose; Gibco Life Technologies, Carlsbad, CA, USA) and supplemented with 10% fetal bovine serum (FBS; Gibco Life Technologies) in a humidified incubator with 95% air and 5% CO2 at 37°C. DMEM without serum was also used in certain experiments. All small hairpin RNA (shRNA)-expressing stable cell lines were grown in DMEM supplemented with 10% tetracycline (Tet)-approved FBS (Clontech Laboratories, Inc., Mountain View, CA, USA) and 1 μg/ml puromycin (Gibco Life Technologies).

Antibodies and agents

The anti-LC3B rabbit polyclonal antibody used in the western blot assay was from Cell Signaling Technology (#12741S; Beverly, MA, USA), the anti-p62/sequestosome 1 (SQSTM1) rabbit polyclonal antibody was obtained from Proteintech (#18420-1-AP; Chicago, IL, USA) and the anti-GAPDH mouse monoclonal antibody was from Beijing CoWin Biotech Co., Ltd. (cw0100A; Beijing, China). Goat anti-rabbit immunoglobulin G-horseradish peroxidase (IgG-HRP) and goat anti-mouse IgG-HRP (Pierce, Thermo Fisher Scientific, Rockford, IL, USA) were used as secondary antibodies, and enhanced chemiluminescence reagent (Pierce, Thermo Fisher Scientific) was used for developing the blots. The agents epirubicin, doxycycline, 3-methyladenine (3-MA; Sigma-Aldrich, St. Louis, MO, USA) and purimycin (Invitrogen Life Technologies, Carlsbad, CA, USA) were dissolved in phosphate-buffered saline (PBS) to form stock solutions and then added directly into the media to the required concentration.

Inducible lentiviral shRNA constructs

To generate shRNA-expressing plasmids, the double-stranded oligonucleotides encoding the desired shRNA were cloned into the AgeI and EcoRI restriction sites of pLKO-Tet-On vector (Addgene, Inc., Cambridge, MA, USA) as described previously (17). The constructs containing an LC3-specific shRNA sequence 5′-CTGAGATCGATCAGTTCAT-3′; and scrambled shRNA 5′-GCAAGCTGACCCTGAAGTTCAT-3′ were designated pLKO-Tet-shLC3 and pLKO-Tet-shCon, respectively.

Virus production and production of stable cell lines

Lentiviruses were generated by co-transfecting 293T cells with 1.5 μg shRNA-encoding plasmid and 1 μg pPAX2 and 0.5 μg pDMG2 (Addgene, Inc.) as helper plasmids, using Lipofectamine 3000 reagent (Invitrogen Life Technologies) according to the manufacturer’s instructions. Growth media was exchanged after 8–16 h and lentivirus-containing supernatant was harvested 24, 48 and 72 h later.

For target cell transduction, HepG2 cells were passaged to 40% confluency the following day. Viral medium was added to the cells with 8 μg/ml Polybrene (Sigma-Aldrich). After 24 h, viral particle-containing medium was removed and replaced with fresh medium containing 1 μg/ml puromycin. From days 4 to 10, fresh medium was replaced when necessary and evaluated for cytotoxicity under a microscope. Finally, the cells, named HepG2shCon and HepG2shLC3, were collected for further experiments.

Immunoblot analysis

Cells were trypsinized, washed with ice-cold PBS and lysed with RIPA buffer (50 mM Tris, 150 mM NaCl, 1% NP-40, 0.5% deoxycholic acid and 0.1% SDS), Protein protease inhibitor mixture (Thermo Fisher Scientific) was added prior to extraction. The lysates were fractionated on an SDS-PAGE gel and transferred onto a Trans-Blot polyvinylidene difluoride membrane (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Blots were blocked with 5% non-fat dry milk followed by probing with the LC3B (1:1,000 dilution, CST) and p62 (1:1,000 dilution, Proteintech) primary antibodies and GAPDH (1:10,000 dilution, CoWin) overnight at 4°C. The membranes were then washed thrice with tyrosylprotein sulfotransferase, incubated with secondary antibodies at room temperature for 2 h and subsequently washed a further three times for developing. The corresponding bands were detected using the Pierce Western HRP protocol.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

RNA was isolated with TRIzol reagent (Invitrogen Life Technologies), treated with DNAse (Promega Corporation, Madison, WI, USA), and then first strand cDNA was created with M-MLV reverse transcriptase (Promega Corporation) according to the manufacturer’s instructions. qPCR was performed in triplicate in 20-μl reactions with iQ SYBR® Premix Ex Taq™ Perfect Real Time (Bio-Rad Laboratories, Inc.), 50 ng first strand cDNA and 0.2 μg each LC3 primer: Forward, 5′-GAGAAGCAGCTTCCTGTTCTGG-3′ and reverse, 5′-GTGTCCGTTCACCAACAGGAAG-3′; or 0.2 g each GAPDH primer: Forward, 5′-GGGTGTGAACCATGAGAAGT-3′ and reverse, 5′-GTAGAGGCAGGGATGATGTT-3′. Samples were cycled once at 95°C for 2 min, then subjected to 40 cycles of 95°C, 58°C and 72°C for 30 sec each. The relative LC3 mRNA content was calculated using the 2−ΔΔCT method with GAPDH as an endogenous control.

Detection of AVOs with acridine orange (AO) staining

Autophagy is characterized by the formation of AVOs. Cells were stained with AO (Sigma-Aldrich) as described previously (18). Briefly, in AO-stained cells, the cytoplasm and nucleolus fluoresce bright green and dim red, respectively, whereas AVOs fluoresce bright red. HepG2shCon and HepG2shLC3 cells were cultured with or without doxycycline (10 μg/ml) for 24 h, then cultured with serum-free DMEM for 2 h. AO was then added at a final concentration of 1 μg/ml for a period of 15 min. The cells were washed with PBS 3 times, and the AVOs were visualized using a fluorescence microscope (Leica DMR, Leica Microsystems GmbH, Wetzlar, Germany) or quantified by flow cytometry (FACScan system; BD Biosciences, San Jose, CA, USA.

Determination of the mean red:green fluorescence ratio in AO-stained cells using flow cytometry

The intensity of the red fluorescence is proportional to the degree of acidity and/or the volume of the cellular acidic compartment. Thus, by comparing the mean red:green fluorescence ratio of different cell populations, the change in the degree of acidity and/or the fractional volume of their cellular acidic compartment was measured. Cells were stained with AO for 15 min, removed from the plate with trypsin-EDTA and collected in phenol red-free growth medium. Green (510–530 nm) and red (>650 nm) fluorescence emissions from 1×104 cells, illuminated with blue (488 nm) excitation light, were measured using a FACScan system and CellQuest software (BD Biosciences).

Cytotoxicity assay

Chemotherapy drug cytotoxicity was assessed in vitro using the Cell Counting kit (CCK)-8 assay (7Sea Biotech, Shanghai, China) as described previously (19). In brief, 2×103 cells were seeded in a 96-well flat-bottomed plate, grown at 37°C for 24 h, and then placed in doxycycline (10 μg/ml). Subsequently, cells were treated with epirubicin at increasing concentrations. After 72 h of culture, 10 μl CCK-8 reagent was added to each well and the plate was incubated for 1 h. Absorbance was read at 450 nm. All samples were carried out in sextuplicate. Data are represented as the percentage reduction in metabolic activity, normalized to HepG2shCon cells.

Cell cycle analysis

Following treatments, cells were trypsinized, washed with PBS and fixed in ice cold 75% ethanol. Cells were then washed with PBS and stained with propidium iodide (Invitrogen Life Technologies) and analyzed by flow cytometry on a FACScan system. Cell cycle distribution was analyzed using CellQuest software.

Statistical analysis

Statistical analysis was performed using GraphPad software (GraphPad Software, Inc., La Jolla, CA, USA). All quantitative data are expressed as the mean ± standard deviation. Statistical differences between groups were compared using a Student’s t-test. P<0.05 was considered to indicate a statistically significant result.

Results

Inducible downregulation of LC3 expression in HepG2 cells

To assess the knockdown of LC3 expression, HepG2shCon and HepG2shLC3 cells were cultured in the presence of increasing amounts of doxycycline (0, 5 and 10 μg/ml) for 72 h and analyzed by immunoblotting and RT-qPCR. In the absence of doxycycline, LC3 protein levels did not differ between the two types of cells. Increasing concentrations of doxycycline, however, resulted in the effective downregulation of LC3 expression in the HepG2shLC3 cells (Fig. 1).

Knockdown of LC3 inhibits serum deprivation-induced autophagy in HepG2 cells

As LC3 has been shown to control the initiation of autophagy, the effect of LC3 inhibition on serum starvation-induced autophagy in HepG2 cells was assessed. AO staining was used to reveal AVOs following serum starvation treatment by fluorescence microscopy and FACS scanning. The data (Fig. 2A and B) demonstrate that the inhibition of LC3 resulted in a marked inhibition of the total number of autophagosomes; the 5 mM 3-MA group served as a positive control. The p62 protein, also known as SQSTM1, is itself degraded by autophagy and may serve to link ubiquitinated proteins to the autophagic machinery to enable their degradation in the lysosome. Therefore, the induction of autophagy after 2 h serum-starvation was assessed using immunoblotting to detect the expression level of p62, with GAPDH as the loading control (Fig. 2C). The degradation of p62 was repressed in the Dox (10 ug/ml) and 3-MA groups. These results indicate that the knockdown of LC3 by inducible shRNA may inhibit starvation-induced autophagy.

Knockdown of LC3 significantly causes cell cycle arrest in HepG2 cells

Epirubicin is a widely used anthracycline drug for chemotherapy that targets DNA by intercalating DNA strands and inhibits DNA and RNA synthesis. The effect of LC3 inhibition on epirubicin-induced cell cycle abnormalities in HepG2 cells was investigated using 5 mM 3-MA treated group as the positive control (Fig. 3). The results indicated that knockdown of LC3 increased the percentage of G1 (2N DNA content) phase cells (Fig. 3C) compared with the shCon group (Fig. 3B) following epirubin treatment. The 3-MA group exhibited a similar result (Fig. 3D) to the shLC3 group.

Silencing of LC3 expression sensitizes HepG2 cells to the therapeutic effect of epirubicin

As autophagy normally promotes the resistance of tumor cells to chemotherapy, whether knockdown of LC3 increases the sensitivity of tumor cells to chemotherapy was investigated. HepG2shCon and HepG2shLC3 cells were treated with epirubicin. The effects of LC3 knockdown with and without epirubicin on cell viability were assessed by CCK-8 assays. LC3 knockdown (induced by 10 μg/ml doxycycline) combined with epirubicin (4 μM) treatment decrease the viability of HepG2shLC3 by ~50% (P<0.01) compared with those of HepG2shLC3 cells treated with epirubicin and HepG2shCon (Fig. 4).

Discussion

HCC is one of the most prevalent cancers worldwide, accounting for 85–90% of all primary liver cancers, which represents ~4% of all newly diagnosed cancer cases (20). Although cytotoxic chemotherapy has been used for >30 years, definite evidence that it prolongs survival has been lacking (21). Various treatment options are available for patients with HCC according to the degree of background liver damage, tumor diameter and other factors associated with disease progression (22). Resistance to chemotherapy drugs remains a significant barrier for cytotoxic agents, often leading to chemotherapy failure in patients with HCC. Understanding of the underlying mechanisms is critical if outcomes are to be improved. There are numerous putative mechanisms for the process by which chemoresistance is induced (2325).

Autophagy was first observed in yeast as a survival mechanism when nutrients were limited, and exists in all eukaryotic cells from yeast to mammals (26). Autophagy is a regulated lysosomal pathway for the degradation and recycling of long-lived proteins and organelles. Nutrient starvation is the most commonly used method for inducing autophagy (27). In autophagy, LC3, a mammalian homolog of yeast ATG8, is activated and relocalizes to intracellular vesicles when the lipid bilayer structure sequesters cytoplasm to form autophagosomes. Firstly, LC3 pro-form is cleaved to form soluble LC3-I. This is then modified to a membrane-bound form, known as LC3-II, which is recruited onto the autophagosomes (28,29). Autophagy occurs in response to various other forms of stress, including oxygen or growth factor deprivation and chemotherapeutics (30,31). Recent evidence suggests that autophagy provides a protective function in tumor cells in response to metabolic stress. A number of studies have reported that autophagy is activated in cancer cells in response to various anticancer therapies (3234). However, little is known about the role of autophagy in HCC chemo-resistance. Epirubicin is a structural analog of doxorubicin that is commonly used to treat HCC. It is usually better tolerated compared with doxorubicin in the treatment of HCC (35).

In the present study, epirubicin-activated autophagy was shown to be inhibited by LC3 RNAi. The observed chemoresistance was found to be associated with the epirubicin-induced activation of autophagy-associated signaling in HepG2 cells. The results show that autophagy can be significantly inhibited by lentivirus-mediated RNAi of LC3 resulting in enhanced cell sensitivity to chemotherapy. These findings suggest that the knockdown of LC3 offers a novel approach that increases the sensitivity of tumor cells to chemotherapeutic agents that target DNA. In addition, they indicate that epirubicin induced autophagy as a pro-survival mechanism in HCC, which caused the HCC cells to be resistant to anthracyclines.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (no. 31100964 and 81372718) and the Specialized Research Fund for Senior Personnel Program of Jiangsu University (no. 10JDG45).

References

1 

Ding J and Wang H: Multiple interactive factors in hepatocarcinogenesis. Cancer Lett. 346:17–23. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Li J, Lau GK, Chen L, Dong SS, Lan HY, et al: Interleukin 17A promotes hepatocellular carcinoma metastasis via NF-κB induced matrix metalloproteinases 2 and 9 expression. PloS One. 6:e218162011. View Article : Google Scholar

3 

Klionsky DJ and Emr SD: Autophagy as a regulated pathway of cellular degradation. Science. 290:1717–1721. 2000. View Article : Google Scholar : PubMed/NCBI

4 

Yoshimori T: Autophagy: a regulated bulk degradation process inside cells. Biochem Biophys Res Commun. 313:453–458. 2004. View Article : Google Scholar

5 

Tanida I, Ueno T and Kominami E: LC3 conjugation system in mammalian autophagy. Int J Biochem Cell Biol. 36:2503–2518. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Okamoto K, Kondo-Okamoto N and Ohsumi Y: Mitochondria-anchored receptor Atg32 mediates degradation of mitochondria via selective autophagy. Dev Cell. 17:87–97. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Reggiori F and Klionsky DJ: Autophagy in the eukaryotic cell. Eukaryotic Cell. 1:11–21. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Turcotte S and Giaccia AJ: Targeting cancer cells through autophagy for anticancer therapy. Curr Opin Cell Biol. 22:246–251. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Hollomon MG, Gordon N, Santiago-O’Farrill JM and Kleinerman ES: Knockdown of autophagy-related protein 5, ATG5, decreases oxidative stress and has an opposing effect on camptothecin-induced cytotoxicity in osteosarcoma cells. BMC Cancer. 13:5002013. View Article : Google Scholar : PubMed/NCBI

10 

Paglin S, Hollister T, Delohery T, Hackett N, McMahill M, et al: A novel response of cancer cells to radiation involves autophagy and formation of acidic vesicles. Cancer Res. 61:439–444. 2001.PubMed/NCBI

11 

Feng Z: p53 regulation of the IGF-1/AKT/mTOR pathways and the endosomal compartment. Cold Spring Harb Perspect Biol. 2:a0010572010. View Article : Google Scholar : PubMed/NCBI

12 

Notte A, Leclere L and Michiels C: Autophagy as a mediator of chemotherapy-induced cell death in cancer. Biochem Pharmacol. 82:427–434. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Huang G, Jiang QH, Cai C, Qu M and Shen W: SCD1 negatively regulates autophagy-induced cell death in human hepatocellular carcinoma through inactivation of the AMPK signaling pathway. Cancer Lett. Dec 17–2014.(Epub ahead of print).

14 

Tian Y, Kuo C, Sir D, et al: Autophagy inhibits oxidative stress and tumor suppressors to exert its dual effect on hepatocarcinogenesis. Cell Death Differ. Dec 19–2014.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

15 

Pan H, Wang Z, Jiang L, et al: Autophagy inhibition sensitizes hepatocellular carcinoma to the multikinase inhibitor linifanib. Sci Rep. 4:66832014. View Article : Google Scholar : PubMed/NCBI

16 

Shi YH, Ding ZB, Zhou J, et al: Targeting autophagy enhances sorafenib lethality for hepatocellular carcinoma via ER stress-related apoptosis. Autophagy. 7:1159–1172. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Wiederschain D, Wee S, Chen L, Loo A, Yang GZ, et al: Single-vector inducible lentiviral RNAi system for oncology target validation. Cell Cycle. 8:498–504. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Paglin S, Hollister T, Delohery T, et al: A novel response of cancer cells to radiation involves autophagy and formation of acidic vesicles. Cancer Res. 61:439–444. 2001.PubMed/NCBI

19 

Yamamoto Y, Mochida J, Sakai D, et al: Upregulation of the viability of nucleus pulposus cells by bone marrow-derived stromal cells: significance of direct cell-to-cell contact in coculture system. Spine (Phila Pa 1976). 29:1508–1514. 2004. View Article : Google Scholar

20 

Jemal A, Bray F, Center MM, et al: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Chen L, Ye HL, Zhang G, Yao WM, Chen XZ, et al: Autophagy inhibition contributes to the synergistic interaction between EGCG and doxorubicin to kill the hepatoma Hep3B cells. PloS One. 9:e857712014. View Article : Google Scholar : PubMed/NCBI

22 

Alberti A, Clumeck N, Collins S, et al; ECC Jury. Short statement of the first European Consensus Conference on the treatment of chronic hepatitis B and C in HIV co-infected patients. J Hepatol. 42:615–624. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Chen J, Ding Z, Peng Y, et al: HIF-1α inhibition reverses multidrug resistance in colon cancer cells via downregulation of MDR1/P-glycoprotein. PLoS One. 9:e988822014. View Article : Google Scholar

24 

Tao W, Shi JF, Zhang Q, Xue B, Sun YJ and Li CJ: Egr-1 enhances drug resistance of breast cancer by modulating MDR1 expression in a GGPPS-independent manner. Biomed Pharmacother. 67:197–202. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Zhou Y, Sun K, Ma Y, et al: Autophagy inhibits chemotherapy-induced apoptosis through downregulating Bad and Bim in hepatocellular carcinoma cells. Sci Rep. 4:53822014.PubMed/NCBI

26 

Meijer AJ and Codogno P: Regulation and role of autophagy in mammalian cells. Int J Biochem Cell Biol. 36:2445–2462. 2004. View Article : Google Scholar : PubMed/NCBI

27 

Li L, Liu GD, Zhang XJ and Li YB: Autophagy, a novel target for chemotherapeutic intervention of thyroid cancer. Cancer Chemother Pharmacol. 73:439–449. 2014. View Article : Google Scholar

28 

Tanida I, Minematsu-Ikeguchi N, Ueno T and Kominami E: Lysosomal turnover, but not a cellular level, of endogenous LC3 is a marker for autophagy. Autophagy. 1:84–91. 2005. View Article : Google Scholar

29 

Kabeya Y, Mizushima N, Ueno T, Yamamoto A, Kirisako T, et al: LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J. 19:5720–5728. 2000. View Article : Google Scholar : PubMed/NCBI

30 

Mizushima N and Levine B: Autophagy in mammalian development and differentiation. Nat Cell Biol. 12:823–830. 2010. View Article : Google Scholar : PubMed/NCBI

31 

White E, Karp C, Strohecker AM, et al: Role of autophagy in suppression of inflammation and cancer. Curr Opin Cell Biol. 22:212–217. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Sui XB, Kong N, Ye L, Han W, Zhou J, et al: p38 and JNK MAPK pathways control the balance of apoptosis and autophagy in response to chemotherapeutic agents. Cancer Lett. 344:174–179. 2014. View Article : Google Scholar

33 

Ge YY, Shi Q, Zheng ZY, et al: MicroRNA-100 promotes the autophagy of hepatocellular carcinoma cells by inhibiting the expression of mTOR and IGF-1R. Oncotarget. 5:6218–6228. 2014.PubMed/NCBI

34 

Yuan H, Li AJ, Ma SL, et al: Inhibition of autophagy significantly enhances combination therapy with sorafenib and HDAC inhibitors for human hepatoma cells. World J Gastroenterol. 20:4953–4962. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Cao H, Phan H and Yang LX: Improved chemotherapy for hepatocellular carcinoma. Anticancer Res. 32:1379–1386. 2012.PubMed/NCBI

Related Articles

Journal Cover

April-2015
Volume 9 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Peng W, Du T, Zhang Z, Du F, Jin J and Gong A: Knockdown of autophagy-related gene LC3 enhances the sensitivity of HepG2 cells to epirubicin. Exp Ther Med 9: 1271-1276, 2015
APA
Peng, W., Du, T., Zhang, Z., Du, F., Jin, J., & Gong, A. (2015). Knockdown of autophagy-related gene LC3 enhances the sensitivity of HepG2 cells to epirubicin. Experimental and Therapeutic Medicine, 9, 1271-1276. https://doi.org/10.3892/etm.2015.2266
MLA
Peng, W., Du, T., Zhang, Z., Du, F., Jin, J., Gong, A."Knockdown of autophagy-related gene LC3 enhances the sensitivity of HepG2 cells to epirubicin". Experimental and Therapeutic Medicine 9.4 (2015): 1271-1276.
Chicago
Peng, W., Du, T., Zhang, Z., Du, F., Jin, J., Gong, A."Knockdown of autophagy-related gene LC3 enhances the sensitivity of HepG2 cells to epirubicin". Experimental and Therapeutic Medicine 9, no. 4 (2015): 1271-1276. https://doi.org/10.3892/etm.2015.2266